MAD2L1 Human

MAD2 Mitotic Arrest Deficient-Like 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of MAD2L1

MAD2L1 (Mitotic Arrest Deficient 2 Like 1), also known as MAD2, is a critical gene located on chromosome 4q27 in humans. It encodes the mitotic spindle assembly checkpoint protein MAD2A, which ensures genomic stability during cell division by delaying anaphase until all chromosomes are properly aligned at the metaphase plate . This checkpoint mechanism prevents chromosomal missegregation, a hallmark of cancer and other genomic instability disorders. MAD2L1 interacts with key regulators like CDC20, BUB1B, and MAD1, forming complexes that inhibit the anaphase-promoting complex/cyclosome (APC/C) until spindle-kinetochore attachment errors are resolved .

Molecular Function and Interactions

MAD2L1 operates in two conformational states: an inactive monomer and an active tetramer. The tetramer binds CDC20 to inhibit APC/C, thereby preventing premature sister chromatid separation . Its interaction network includes:

  • CDC20: Critical for APC/C activation .

  • BUB1B: Cooperates in spindle checkpoint signaling .

  • MAD1: Stabilizes MAD2L1 at unattached kinetochores .

  • UBD (Ubiquitin D): Implicated in checkpoint regulation .

Key pathways:

  • Mitotic spindle checkpoint signaling (GO:0007094) .

  • Regulation of anaphase-promoting complex (APC/C) .

Role in Cancer

MAD2L1 dysregulation is strongly linked to oncogenesis:

Cancer TypeFindingsReferences
Breast CancerHeterozygous frameshift mutations (e.g., 572 del A) observed in cell lines; altered expression correlates with aneuploidy and chromosomal instability .
Lung AdenocarcinomaHigh MAD2L1 expression predicts poor prognosis (HR = 1.8 for OS, p = 0.01) .
Liver CancerMad2l1/Trp53 double-knockout mice develop exponential tumor growth, mimicking human hepatocellular carcinoma .
MesotheliomaMAD2L1 identified as a driver gene; inhibitor Neratinib shows therapeutic potential .

Pulmonary Fibrosis

MAD2L1 is upregulated in alveolar epithelial cells of idiopathic pulmonary fibrosis (IPF) patients. Inhibition exacerbates mitochondrial damage, ROS production, and senescence, promoting fibrosis .

Cellular Senescence

MAD2L1 depletion triggers premature senescence in fibroblasts via p53 activation, linking checkpoint failure to aging-related processes .

Genetic and Clinical Insights

  • Polymorphisms: Maternal MAD2L1 Leu84Met (rs1283639804) is associated with fetal chromosomal abnormalities .

  • Expression Variants: Reduced MAD2L1 levels correlate with chemoresistance in ovarian cancer .

Clinical Implications and Therapeutic Potential

  • Biomarker Potential: MAD2L1 overexpression in lung and breast cancers serves as a prognostic marker .

  • Therapeutic Targets: Inhibitors like Neratinib (targeting MAD2L1) show efficacy in preclinical models .

Product Specs

Introduction
MAD2L1-binding protein (MAD2L1) is a key player in the mitotic spindle assembly checkpoint, ensuring that anaphase doesn't begin prematurely until all chromosomes are correctly positioned at the metaphase plate. This protein is crucial for the mitotic checkpoint's function, which involves monitoring the connection between kinetochores and spindle fibers. By sequestering CDC20, MAD2L1 inhibits the anaphase promoting complex until all chromosomes are properly aligned.
Description
MAD2L1 Human Recombinant, produced in E.Coli, is a single, non-glycosylated polypeptide chain consisting of 225 amino acids (1-205 a.a.) with a molecular weight of 25.7kDa. This protein is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
MAD2L1 protein solution at a concentration of 0.5mg/ml in a buffer containing 20mM Tris-HCl (pH 8.0), 20% glycerol, 0.1M NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms

Mitotic spindle assembly checkpoint protein MAD2A, HsMAD2, Mitotic arrest deficient 2-like protein 1, MAD2-like protein 1, MAD2L1, MAD2.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MALQLSREQG ITLRGSAEIV AEFFSFGINS ILYQRGIYPS ETFTRVQKYG LTLLVTTDLE LIKYLNNVVE QLKDWLYKCS VQKLVVVISN IESGEVLERW QFDIECDKTA KDDSAPREKS QKAIQDEIRS VIRQITATVT FLPLLEVSCS FDLLIYTDKD LVVPEKWEES GPQFITNSEE VRLRSFTTTI HKVNSMVAYK IPVND.

Q&A

What is MAD2L1 and what is its primary function in human cells?

MAD2L1 (Mitotic Arrest Deficient 2 Like 1) is a critical component of the spindle assembly checkpoint (SAC) that prevents the onset of anaphase until all chromosomes are properly aligned at the metaphase plate during cell division. It functions in an interactive manner with other SAC proteins to initiate checkpoint signals that prevent premature chromosome segregation, thereby safeguarding genomic stability. The protein plays a fundamental role in preventing aneuploidy, which is an abnormal number of chromosomes in daughter cells. This checkpoint mechanism is essential for maintaining the fidelity of chromosome distribution during mitosis and preserving genomic integrity across cell generations .

How does MAD2L1 dysregulation contribute to disease pathogenesis?

Dysregulation of MAD2L1 is implicated in multiple disease processes, primarily through its effects on chromosomal stability. When MAD2L1 function is compromised, cells may undergo improper chromosome segregation, leading to aneuploidy and chromosomal instability (CIN). This genomic instability can promote tumorigenesis in various tissues. Research indicates that MAD2L1 aberrations contribute to colorectal cancer susceptibility, with specific genetic variants like Arg558His showing significant associations with cancer risk (OR = 1.38, 95% CI: 1.09–1.75) . In liver tissue, MAD2L1 overexpression supports MYC-driven hepatocarcinogenesis, where silencing MAD2L1 reduces cell growth in vitro and inhibits cancer development in vivo . Additionally, in pulmonary tissues, inhibition of MAD2L1 impairs mitochondrial function and induces cell senescence, potentially promoting the establishment of a profibrotic microenvironment that contributes to pulmonary fibrosis .

What clinical significance does MAD2L1 expression have in cancer diagnostics?

MAD2L1 expression shows significant potential as a diagnostic biomarker, particularly in breast cancer. Studies examining MAD2L1 expression in breast cancer tissues have demonstrated its overexpression compared to normal tissues, with this upregulation significantly associated with higher clinical stage and histological grade of breast cancer. Statistical analysis reveals a strong diagnostic capacity, with an area under curve (AUC) value of 0.9642 from SROC (summarized receiver operating characteristic) curve analysis . Furthermore, significant correlations have been established between MAD2L1 expression and several established tumor indicators including estrogen receptor (ER), P53, HER-2, and Ki-67, enhancing its utility in comprehensive diagnostic panels . These findings suggest that MAD2L1 detection could substantially improve the diagnosis and prognostic evaluation of breast cancer, potentially serving as part of a molecular signature for cancer classification and treatment stratification.

How does MAD2L1 contribute to different cancer types?

MAD2L1 exhibits tissue-specific roles in carcinogenesis across multiple cancer types. In colorectal cancer (CRC), genetic variants in MAD2L1 (particularly Leu84Met) significantly enhance cancer risk, especially when interacting with environmental factors like smoking (P = 0.016) . For hepatocellular carcinoma (HCC), MAD2L1 is consistently upregulated in tumor tissues and associated with higher aggressive tumor grade and elevated proliferative activity. RNA-seq results from HCC cell lines demonstrate that MAD2L1 silencing induces the expression of genes associated with cell cycle regulation, DNA replication, and various cancer-related pathways, highlighting its critical role in liver cancer progression . In breast cancer, MAD2L1 overexpression correlates with higher clinical stage and histological grade, and significant associations have been found between MAD2L1 expression and tumor indicators including ER, P53, HER-2, and Ki-67 . In T-cell lymphomas, MAD2L1 deletion combined with Trp53 mutation leads to rapidly growing acute lymphoblastic leukemia (T-ALL) characterized by ongoing chromosomal instability but with tissue-specific patterns of chromosome gain and loss .

What are the molecular mechanisms by which MAD2L1 variants influence cancer risk?

MAD2L1 variants influence cancer risk through multiple molecular mechanisms centered on chromosomal stability regulation. The Leu84Met variant of MAD2L1 has been shown to significantly interact with environmental factors such as smoking to enhance colorectal cancer risk (P = 0.016), suggesting altered protein-environment interactions . This variant may impair the protein's ability to properly function in the spindle assembly checkpoint. Mechanistically, MAD2L1 variants can compromise the protein's ability to bind to other SAC components, reducing the efficiency of the mitotic checkpoint and allowing cells with misaligned chromosomes to progress through mitosis. In breast cancer research, one heterozygous frameshift mutation (572 del A) has been identified that creates a truncated MAD2 protein product, potentially compromising its function . Additionally, genomic analyses of tumors with MAD2L1 abnormalities reveal recurrent and tissue-specific patterns of chromosome loss and gain, suggesting that while MAD2L1 dysfunction causes ongoing chromosomal instability, specific aneuploid configurations may be selected based on tissue context and microenvironment, ultimately driving the oncogenic process in a tissue-specific manner .

What is the prognostic value of MAD2L1 expression in different cancers?

MAD2L1 expression serves as a significant prognostic indicator across multiple cancer types. In hepatocellular carcinoma (HCC), elevated MAD2L1 expression is strongly associated with poor prognosis, higher aggressive tumor grade, and increased proliferative activity . This association with aggressive disease features makes MAD2L1 a valuable marker for risk stratification in liver cancer patients. In breast cancer, comprehensive analysis of MAD2L1 expression using immunohistochemistry in 209 invasive ductal breast cancer samples, combined with RNA-sequencing and microarray data from multiple databases, has established its overexpression as significantly correlated with higher clinical stage and histological grade . The relationship between MAD2L1 expression and established prognostic markers (ER, P53, HER-2, and Ki-67) further underscores its utility in building comprehensive prognostic models. Additionally, in colorectal cancer, specific MAD2L1 variants (such as Arg558His) correlate with increased chromosomal instability as measured by micronucleus assays, potentially serving as biomarkers for identifying patients at higher risk for aggressive disease progression . These findings collectively suggest that MAD2L1 expression analysis could significantly enhance current prognostic assessment methods in clinical oncology.

What are the preferred methods for studying MAD2L1 expression in clinical samples?

Several complementary methods are recommended for comprehensive analysis of MAD2L1 expression in clinical samples. Immunohistochemistry (IHC) represents a standard approach for protein-level detection in tissue samples, as demonstrated in studies examining MAD2L1 expression in 209 invasive ductal breast cancer samples . This technique allows for spatial visualization of protein expression within the tumor microenvironment. At the transcript level, quantitative real-time PCR (qRT-PCR) provides sensitive detection of MAD2L1 mRNA expression, as employed in studies of both pulmonary fibrosis tissues and cancer samples . For broader expression profiling, RNA-sequencing offers comprehensive insights into MAD2L1 expression patterns in relation to other genes, particularly useful for pathway analysis as shown in MAD2L1 knockdown studies in HCC cell lines . Western blot analysis complements these approaches by providing semi-quantitative assessment of protein levels and can detect specific protein variants, as utilized in comparative studies of normal versus fibrotic lung tissues . Additionally, microarray-based techniques enable high-throughput analysis of expression patterns across large sample cohorts, as demonstrated in meta-analyses of Gene Expression Omnibus (GEO) datasets comprising 663 breast cancer samples and 289 normal samples .

How can researchers effectively model MAD2L1 dysfunction in experimental systems?

Researchers can employ several complementary approaches to model MAD2L1 dysfunction in experimental systems. CRISPR-Cas9 genome editing represents a powerful tool for generating conditional knockout models, as demonstrated in studies where Mad2l1 was specifically silenced in c-MYC-induced mouse HCC models, effectively preventing cancer development . Conditional gene deletion using tissue-specific promoters (such as Alb-Cre for hepatocyte-specific deletion or T-cell specific promoters) allows investigation of MAD2L1 loss in specific tissues while avoiding embryonic lethality that might result from complete knockout . RNA interference (RNAi) approaches using siRNA or shRNA provide flexibility for temporary and partial knockdown of MAD2L1 expression, particularly useful in cell line studies where complete deletion might be lethal. For example, MAD2L1 knockdown in human HCC cell lines has revealed its role in regulating cell growth . Small molecule inhibitors targeting the protein's function rather than expression can also model dysfunction without eliminating the protein. Additionally, introducing specific point mutations that mimic human variants (such as the Leu84Met variant) through site-directed mutagenesis allows investigation of how specific alterations affect protein function . Importantly, when modeling checkpoint dysfunction, researchers should consider combining MAD2L1 manipulation with alteration of other genes like Trp53, as this combination more accurately recapitulates human disease conditions where multiple genetic abnormalities coexist .

What techniques are most effective for assessing chromosomal instability resulting from MAD2L1 dysfunction?

Multiple complementary techniques are recommended for comprehensive assessment of chromosomal instability (CIN) resulting from MAD2L1 dysfunction. The cytokinesis-block micronucleus cytome assay represents a validated approach for quantifying chromosomal damage, as employed in studies examining the effect of MAD2L1 variants on CIN . This technique allows visualization and enumeration of micronuclei in binucleated cells, providing a direct measure of chromosome missegregation events. Single-cell sequencing offers unprecedented resolution for detecting cell-to-cell variation in chromosome copy number, revealing elevated rates of chromosome missegregation in MAD2L1-null T-cell acute lymphoblastic leukemias compared to normal T-cells . For population-level assessment, array-based comparative genomic hybridization (aCGH) effectively identifies recurrent patterns of chromosome loss and gain across tumor samples, demonstrating tissue-specific aneuploid patterns in MAD2L1-null tumors . Fluorescence in situ hybridization (FISH) using chromosome-specific probes provides another approach for visualizing and quantifying specific chromosome gains or losses. Additionally, time-lapse microscopy of cells expressing fluorescent markers for chromosomes or kinetochores enables real-time visualization of chromosome missegregation events during mitosis in MAD2L1-deficient cells. The integration of these techniques provides a comprehensive view of both the frequency and specific patterns of chromosomal abnormalities resulting from MAD2L1 dysfunction across different experimental systems.

How do gene-environment interactions influence MAD2L1-related cancer risk?

Gene-environment interactions significantly modulate MAD2L1-related cancer risk, with smoking emerging as a particularly important environmental factor. Research has demonstrated significant multiplicative gene-smoking interactions with MAD2L1 variants, specifically Arg558His (P = 0.019) and Leu84Met (P = 0.016), that substantially enhance colorectal cancer risk . These findings suggest that environmental exposures may exacerbate the genomic instability resulting from MAD2L1 dysfunction. Mechanistically, carcinogens in tobacco smoke might induce DNA damage that, when combined with compromised spindle assembly checkpoint function due to MAD2L1 variants, leads to amplified chromosomal instability and accelerated carcinogenesis. This is supported by observations that the frequencies of lymphocytic micro-nucleated binucleated cells for MAD2L1 Arg558His polymorphism were significantly different in smoking-exposed groups (P = 0.013) but not in control groups . These interactions likely involve complex cellular responses where environmental stressors trigger compensatory mechanisms that become maladaptive in the context of MAD2L1 dysfunction. Understanding these interactions requires integrative approaches that combine genetic analysis, exposure assessment, and functional studies to elucidate how specific environmental factors influence the cellular consequences of MAD2L1 variants, potentially identifying high-risk populations that might benefit from targeted screening or intervention strategies.

What is the relationship between MAD2L1 expression and mitochondrial function in disease contexts?

MAD2L1 demonstrates a complex relationship with mitochondrial function that appears particularly relevant in pulmonary fibrosis pathogenesis. Research has revealed that inhibition of MAD2L1 results in mitochondrial damage characterized by altered membrane potential and compromised respiratory function . This mitochondrial dysfunction leads to augmented reactive oxygen species (ROS) production, triggering cellular senescence pathways that promote the establishment of a profibrotic microenvironment in lung tissue. The mechanistic connection between MAD2L1, a canonical spindle checkpoint protein, and mitochondrial function remains to be fully elucidated, but may involve indirect regulation of mitochondrial quality control pathways or mitochondrial dynamics. The relationship is likely bidirectional, as mitochondrial dysfunction and resulting ROS can induce DNA damage that may further compromise chromosomal stability in cells with MAD2L1 abnormalities. This creates a potential feedback loop where initial MAD2L1 dysfunction leads to mitochondrial impairment, which further exacerbates genomic instability. These findings suggest that therapeutic strategies targeting mitochondrial function, such as antioxidants or mitochondrial-targeted compounds, might mitigate disease progression in contexts where MAD2L1 dysfunction contributes to pathology . Further research is needed to characterize the molecular intermediaries connecting MAD2L1 status to mitochondrial health across different tissue contexts and disease states.

How does MAD2L1 interact with other cancer-related pathways such as MYC signaling?

MAD2L1 exhibits significant functional interactions with major oncogenic pathways, particularly the MYC signaling network. In hepatocellular carcinoma models, MAD2L1 expression is markedly upregulated in c-MYC-induced tumors, suggesting co-regulation or functional cooperation between these genes . Critically, CRISPR-mediated silencing of Mad2l1 prevented c-MYC-driven mouse liver cancer development, establishing MAD2L1 as an essential downstream effector or facilitator of MYC-induced oncogenesis . RNA-sequencing analysis of MAD2L1 knockdown in HCC cell lines reveals altered expression of genes associated with cell cycle regulation, DNA replication, and various cancer-related pathways, indicating broad transcriptional effects beyond its canonical mitotic checkpoint function . The interaction likely involves both direct and indirect mechanisms, where MAD2L1 may facilitate MYC-driven proliferation by enabling rapid cell division while tolerating the genomic instability that typically results from accelerated replication. This relationship appears to be tissue-specific, as demonstrated by the distinctive patterns of chromosomal gains and losses observed in different MAD2L1-deficient tumor types . In contexts where both MAD2L1 and tumor suppressor pathways like p53 are dysregulated, as in Alb-Cre::Mad2l1f/f::Trp53R246S mice, particularly aggressive disease phenotypes emerge, highlighting the importance of considering pathway interactions in multiple-hit models of carcinogenesis . These findings suggest that therapeutic strategies targeting MAD2L1 might be particularly effective in MYC-driven cancers, potentially disrupting this oncogenic cooperation.

How can we reconcile contradictory findings regarding MAD2L1 expression across different cancer types?

The contradictory findings regarding MAD2L1 expression across cancer types reflect the context-dependent nature of its function in tumorigenesis. While some studies report MAD2L1 overexpression in breast cancer and hepatocellular carcinoma , others have documented decreased expression in certain breast cancer cell lines exhibiting chromosomal instability . These apparent contradictions can be reconciled by considering several factors: First, the stage-specific expression patterns suggest that MAD2L1 may be upregulated in early carcinogenesis to facilitate rapid proliferation but may be downregulated in later stages in some tumors as they develop tolerance to aneuploidy. Second, tissue-specific regulatory mechanisms likely influence how MAD2L1 dysregulation affects cell fitness in different cellular contexts. Third, methodological differences in expression analysis (mRNA vs. protein, bulk tissue vs. cellular resolution) may account for some discrepancies between studies. Fourth, genetic background effects, particularly the status of related checkpoint genes and p53, significantly influence how MAD2L1 abnormalities manifest phenotypically. This is evident in mouse models where Mad2l1 deletion combined with Trp53 mutation produces distinctive tissue-specific cancer phenotypes . Additionally, population-level genomic analyses often mask the heterogeneity revealed by single-cell approaches, as demonstrated by the difference between single-cell sequencing showing ongoing chromosomal instability and array-CGH showing recurrent patterns of aneuploidy in MAD2L1-null tumors . Future research should employ multiple complementary approaches across well-defined disease stages and genetic backgrounds to develop a unified model of MAD2L1's role in cancer.

What are the current limitations in translating MAD2L1 research into clinical applications?

Despite significant advances in understanding MAD2L1 biology, several limitations impede translation into clinical applications. First, the complex context-dependency of MAD2L1 function across different tissues and genetic backgrounds complicates the development of broadly applicable therapeutic approaches. The observation that MAD2L1-null tumors display tissue-specific patterns of aneuploidy suggests that targeting strategies may need to be customized for different cancer types . Second, the essential nature of the spindle assembly checkpoint in normal dividing cells creates a narrow therapeutic window for direct MAD2L1 targeting, raising concerns about potential toxicity to rapidly dividing normal tissues. Third, inconsistent methodologies for assessing MAD2L1 status across studies (ranging from immunohistochemistry to RNA expression to genetic variants) make it difficult to establish standardized clinical assays with validated cutoff values for diagnostic or prognostic applications . Fourth, while genetic association studies have identified MAD2L1 variants associated with cancer risk, the functional consequences of many variants remain poorly characterized, limiting their utility as predictive biomarkers . Fifth, the relationship between MAD2L1 and established cancer therapies, particularly those targeting cell division, remains underexplored, creating uncertainty about how MAD2L1 status might influence treatment response. Finally, the development of specific MAD2L1 modulators faces significant pharmaceutical challenges, as protein-protein interactions critical to checkpoint function can be difficult to target with small molecules. Addressing these limitations will require integrated approaches combining mechanistic studies, biomarker validation in large patient cohorts, and creative drug development strategies that exploit synthetic lethal interactions with MAD2L1 dysfunction.

Table 1: MAD2L1 Genetic Variants Associated with Cancer Risk

Genetic VariantCancer TypeOdds Ratio (95% CI)P-valueReference
MAD2L1 Arg558HisColorectal Cancer1.38 (1.09-1.75)<0.05
MAD2L1 Leu84MetColorectal Cancer1.48 (1.09-2.01)<0.05
572 del A (frameshift)Breast CancerNot quantifiedNot reported

Table 2: Expression Patterns of MAD2L1 Across Cancer Types

Cancer TypeExpression PatternAssociation with Clinical FeaturesPrognostic SignificanceReference
Hepatocellular CarcinomaUpregulatedHigher aggressive tumor grade, elevated proliferative activityPoor prognosis
Breast CancerOverexpressedHigher clinical stage, histological gradePotential diagnostic value (AUC = 0.9642)
Colorectal CancerNot specifiedAssociated with chromosomal instabilityNot reported
T-cell LymphomaDeletion (with Trp53 mutation)Rapid growth in T-ALLNot reported

Table 3: MAD2L1 Interactions with Environmental and Genetic Factors

Interaction FactorEffectStatistical SignificanceCancer TypeReference
Smoking × MAD2L1 Arg558HisEnhanced cancer riskP = 0.019Colorectal Cancer
Smoking × MAD2L1 Leu84MetEnhanced cancer riskP = 0.016Colorectal Cancer
c-MYCMAD2L1 required for MYC-driven carcinogenesisSignificant prevention of tumor developmentHepatocellular Carcinoma
Trp53 mutationAccelerated and aggressive cancer developmentP < 0.001Hepatocellular Carcinoma

Product Science Overview

Discovery and Cloning

The human orthologues of MAD2, namely MAD2L1 and MAD2L2, were first cloned in a search for human complementary DNAs (cDNAs) that could rescue the microtubule poison-sensitivity of a yeast strain lacking a kinetochore binding protein . This discovery highlighted the importance of MAD2L1 in the spindle assembly checkpoint and its evolutionary conservation across species.

Function and Mechanism

MAD2L1 functions as a component of the spindle-assembly checkpoint by forming a heterotetrameric complex with MAD1L1 at unattached kinetochores during prometaphase. This complex recruits open conformation molecules of MAD2L1 (O-MAD2) and promotes their conversion to the closed conformation (C-MAD2). The C-MAD2 then sequesters CDC20, inhibiting the activity of the anaphase-promoting complex (APC/C) until all chromosomes are properly aligned .

Role in Disease

Mutations and dysregulation of MAD2L1 have been associated with various diseases, including developmental and epileptic encephalopathy 52 and gastric cancer . Additionally, genetic variants in MAD2L1 have been linked to an increased risk of lung cancer, as they can impair spindle checkpoint function, leading to chromosomal instability .

Research and Applications

Research on MAD2L1 continues to be of great interest due to its critical role in maintaining genomic stability. Studies have focused on understanding the functional relevance of missense variations in MAD2L1 and their impact on cancer susceptibility . The recombinant form of MAD2L1 is used in various experimental setups to study its function and interactions with other proteins involved in the mitotic checkpoint.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.